US4684623A - Use of tumor necrosis factor as a weight regulator - Google Patents

Use of tumor necrosis factor as a weight regulator Download PDF

Info

Publication number
US4684623A
US4684623A US06/801,989 US80198985A US4684623A US 4684623 A US4684623 A US 4684623A US 80198985 A US80198985 A US 80198985A US 4684623 A US4684623 A US 4684623A
Authority
US
United States
Prior art keywords
tnf
ser
val
2ser
10ser
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US06/801,989
Inventor
James W. Larrick
Gordon M. Ringold
David F. Mark
Leo S. Lin
Frank M. Torti
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis Vaccines and Diagnostics Inc
Leland Stanford Junior University
Original Assignee
Cetus Corp
Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US06/801,989 priority Critical patent/US4684623A/en
Application filed by Cetus Corp, Leland Stanford Junior University filed Critical Cetus Corp
Assigned to CETUS CORPORATION, A CORP OF DELAWARE reassignment CETUS CORPORATION, A CORP OF DELAWARE ASSIGNMENT OF ASSIGNORS INTEREST. Assignors: LARRICK, JAMES W., LIN, LEO S., MARK, DAVID F.
Assigned to BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY THE, A CORP OF CALIFORNIA reassignment BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY THE, A CORP OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST. Assignors: RINGOLD, GORDON M., TORTI, FRANK M.
Priority to PCT/US1986/000952 priority patent/WO1986006280A1/en
Priority to EP86903084A priority patent/EP0226603A1/en
Priority to AU58628/86A priority patent/AU5862886A/en
Priority to FI865373A priority patent/FI865373A0/en
Priority to DK001887A priority patent/DK1887A/en
Priority to NO870008A priority patent/NO870008L/en
Publication of US4684623A publication Critical patent/US4684623A/en
Application granted granted Critical
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S930/00Peptide or protein sequence
    • Y10S930/01Peptide or protein sequence
    • Y10S930/14Lymphokine; related peptides
    • Y10S930/144Tumor necrosis factor

Definitions

  • the invention relates to control of lipid metabolism using tumor necrosis factor (TNF). Specifically, the invention relates to a method of controlling obesity by administration of TNF.
  • TNF tumor necrosis factor
  • cachectin A crude protein extract from the media of endotoxin-stimulated macrophages, designated "cachectin", has been shown to induce indicia of cachexia in tissue culture (Torti, F., et al., Science (1985) 229: 867-869), and antibodies raised against it have been shown to protect mice from some of the effects of E. coli lipopolysaccharide endotoxin (Beutler, B., et al., ibid, pp. 869-871).
  • TNF tumor necrosis factor
  • the invention provides a defined pharmaceutical, tumor necrosis factor (TNF), which is capable of stimulating the physiological state associated with cachexia.
  • TNF tumor necrosis factor
  • the control of weight in adipose persons is thereby effected using a material which simulates a natural physiological response to infection under conditions where such simulation can be regulated and controlled.
  • the invention relates to a method for controlling obesity by administration of TNF.
  • the invention relates to controlling cachexia in cancer or infectious disease patients by providing antibodies to neutralize TNF.
  • the invention relates to pharmaceutical compositions useful for weight reduction which contain TNF as an active ingredient, and to immunoglobulin compositions useful in neutralizing TNF.
  • FIG. 1 shows the nucleotide sequence encoding human mature TNF and the deduced amino acid sequence.
  • FIG. 2 shows the accumulation of adipose-inducible mRNAs in maturing adipose cells and the regulation of synthesis of such mRNA in the presence and absence of TNF.
  • FIG. 3 shows the depression of expression of adipose stimulated genes by TNF.
  • FIGS. 4A and 4B are photographs of mature adipose cells with and without TNF.
  • tumor necrosis factor refers to an amino acid sequence typified by that shown in FIG. 1, which is capable of selective cytotoxicity against tumor cells.
  • the sequence of FIG. 1 is derived from a human cDNA, but TNF encoded by other mammalian species may exhibit the required activity as well. Retrieval and deduction of this sequence is described in detail in U.S. Ser. No. 760,661, filed July 30, 1985, assigned to the same assignee, and incorporated herein by reference. (See also, Wang, et al., Science (1985) 228:149.)
  • TNF amino acid sequence to fit the definition herein, must be active in the in vitro cytotoxicity assay based on the continuous murine connective tissue cell line L-929 as described hereinbelow. It is recognized that this definition of TNF activity is not precisely the same as that set forth in the disclosure coining this term by Carswell, et al. Proc Natl Acad Sci (USA) (1975) 72:3666. However, this activity as confirmed by the in vitro cytotoxicity assay against human tumor cells provides sufficient assurance of utility that qualification as a tumor necrosis factor for humans using this assay is justified; cytotoxicity against L-929 generalizes to human tumors. It is also expected that there is a substantial overlap between factors active in the specified cytotoxicity assay and the in vivo assay outlined by Carswell.
  • the TNF protein of the invention may be in the form of pharmaceutically acceptable salts or may be in neutral form.
  • the free amino groups of the protein are, of course, capable of forming acid addition salts with, for example, inorganic acids such as hydrochloric, phosphoric, or sulfuric acid; or with organic acids such as, for example, acetic glycolic, succinic, or mandelic acid.
  • the free carboxyl groups are capable of forming salts with bases, including inorganic bases such as sodium, potassium, or calcium hydroxides, and such organic bases as piperidine, glucosamine, trimethylamine, chloine, and caffeine.
  • the protein may be modified by combination with other biological materials such as lipids and saccharides, or by side chain modification such as acetylation of amino groups, phosphorylation of hydroxyl side chains, or oxidation of sulfhydryl groups. All of these modifications are included within the scope of the definition, so long as the TNF activity is retained.
  • mutein lacking the first four amino acids at the N-terminus of the sequence shown in FIG. 1 has a specific activity several fold higher than the TNF of the structure shown.
  • TNF specifically includes this truncated form.
  • muteins lacking the N-terminal ten or less amino acids have similarly enhanced activity, and it appears that deletions of up to 10 amino acids from the N-terminus do not destroy, but, in fact, enhance biological activity.
  • TNF specifically includes proteins having an amino acid sequence substantially equivalent to that shown in FIG. 1, but lacking 1-10 of the amino acids at the N-terminal sequence as shown in that figure.
  • muteins of mTNF and will be denoted as to their differences from mTNF using the numbering of residues shown in the figure.
  • muteins which have substitutions for cysteine at position 69 will be denoted using the substituted residue and the position number, e.g., peptides having a serine in place of the cysteine at position 69 are designated ser 69 TNF.
  • Muteins which lack, for example, three N-terminal amino acids as compared to the protein shown in FIG. 1 will be designated ⁇ 3TNF. Where both of the foregoing alterations are made, the mutein is designated ⁇ 3ser 69 TNF.
  • TNF of the inventions include ⁇ 2ser 69 TNF, ⁇ 2ser 101 TNF, ⁇ 2ser 69 ser 101 TNF, and the corresponding ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9 and ⁇ 10 cysteine depleted muteins.
  • Particularly preferred are ⁇ 8ser 69 TNF, ⁇ 8ser 101 TNF, ⁇ 8ser 69 ser 101 and ⁇ 4ser 69 TNF, ⁇ 4ser 101 TNF and ⁇ 4ser 69 ser 101 TNF.
  • FIG. 1 has an additional pair of serine residues following the serine at position 3 as compared to HL-60 derived TNF before resuming the homology shown between positions 4-12 of the HL-60 derived protein and positions 6-14 of the deduced sequence.
  • positions 13 and 14 of the HL-60 derived protein are Val-Ser; the corresponding positions 15 and 16 of the deduced sequence are His-Val.
  • the TNF useful in the method of the invention can most conveniently be prepared using recombinant methods.
  • Detailed descriptions of ways to produce recombinant TNF are set forth in U.S. Ser. No. 661,026 Oct. 15, 1984), Ser. No. 670,360 (Sept. 11, 1984), Ser. No. 698,939 (Feb. 7, 1985), Ser. No. 730,696 ( May 2, 1985), and Ser. No. 760,661 (supra), all incorporated by reference.
  • various DNA sequences enconding TNF have been deposited with the American Type Culture Collection, Rockville, MD.
  • DNA sequences include those contained in pE4 which harbors the human cDNA insert (ATCC #39894); pAW711 which is an expression vector suitable for procaryotes encoding the TNF of FIG. 1 (ATCC #39918) and pAW736, an expression vector encoding the ⁇ 4 mutein of mTNF (ATCC #53092).
  • Vectors suitable for expression of other TNF muteins are deposited as ATCC Nos. 53161, 53162, 53163, 53164, and 53165, respectively.
  • the TNF may be extracted from natural source such as human or other mammalian tissues, or from human or other mammalian derived cell lines.
  • the source of the protein is, of course, immaterial to the practice of the invention except as it may affect specific dosage levels and administration regimes required.
  • the active ingredient may be formulated using a variety of acceptable excipients as is known in the art.
  • the TNF will be administered by injection, either intraperitoneally or intravenously.
  • suitable formulations it may be possible to obtain compositions which may be topically or orally administered, or which may be capable of transmission across mucous membranes.
  • compositions may be in solid or liquid form.
  • conventional carriers include, for example, phamaceutical grades of mannitol, lactose, starch, talcum, cellulose, magnesium carbonate and the like.
  • the TNF may be formulated as a suppository using, for example, polyalkylene glycols as carrier.
  • Liquid compositions can be prepared by dissolving or dispersing the TNF and optional adjuvants in a carrier such as, for example, water, saline, aqueous dextrose, and so forth.
  • composition may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and the like. Actual methods of preparing dosage forms are known or will be apparent to those of the art.
  • the composition will, in any event, contain a quantity of TNF in an amount effective to effect the desired lipid mobilization.
  • a preferred method of administration of this protein is by injection, most commonly intraperitoneally or intravenously.
  • the injectable can be prepared either as a liquid solution or a suspension or in a solid form susceptible to reconstitution.
  • the dosage and mode of administration will, of course, depend on the level of lipid mobilization desired, the nature of the subject, and the judgment of the medical practitioner. In general, however, effective dosages are in the range of 0.02-2 ⁇ g of TNF per kg of body weight per day for so long as administration is required. This range represents, of course, a broad estimate as the above-listed factors are of great importance in determining optimal dosage levels.
  • a preferred embodiment to neutralize the activity of TNF employs polyclonal or monoclonal antibodies. These neutralizing antibodies are preferably used as adjunctive treatment with other cytotoxic drugs in cancer patients.
  • the patient might be administered methotrexate to kill the tumor followed by the antibody preparation to neutralize TNF to prevent weight loss.
  • Polyclonal antibodies are prepared to TNF using conventional procedures by injecting purified TNF into a convenient host, such as rabbits or mice, and harvesting high titer sera. Preparation of monoclonal compositions generally follows the now well known procedure of Kohler and Milstein.
  • Cachectin was used as a control in the following examples. It is prepared from the conditioned media of endotoxin stimulated macrophages, as described by Kawakami, M., et al., Proc Natl Acad Sci (USA) (1982) 79:912; Pekala, P. H., et al., (ibid 1983) 80:2743.
  • TNF prepared by culturing pAW711 transformed E. coli and inducing the expression of TNF as described in Ser. No. 760,661 (supra) was added to the cell cultures from preconfluence to 6 days thereafter.
  • TAl cells which are derived from 5-azacytidine treatment of 10 T1/2C18 cells (Resnikof, C., et al., Cancer Res (1973) 33:3231-3238; Taylor, S. M., et al., Cell (1979) 17:771-779) were grown in Eagle's basal medium supplemented with 10% heat inactivated fetal calf serum. 10 -6 M dexamethasone was present in media for the first 3 days after confluence, and 5 ⁇ g/ml bovine insulin for the first 6 days after confluence.
  • Recombinant TNF (10-30 ng/ml) was first added to preadipocyte cultures 2 days prior to confluence. (Addition of this amount inhibits 90% of lipoprotein lipase activity in cultured adipocytes.) Cells were fed with resupplementation of TNF at day 0 (confluence) and at day 3. Cells were harvested at day 6.
  • Nick translated cDNA clones of adipose-inducible genes (designated clones 1, 10, 47, and glycerol phosphate dehydrogenase (GDH)) were used to probe the filters under hybridization conditions described in Chapman, A. B., et al., J Biol Chem (1984) (supra). Filters were washed and exposed to XAR 5 film at -70° C. with an intensifying screen.
  • Lipid accumulation was also completely inhibited by cachectin or TNF, and TAl cell cultures treated with these compounds have been maintained for as long as 23 days without the appearance of neutral lipid evident by oil red O staining.
  • adipocyte morphology returns, as does the expression of adipose-inducible genes. It was also shown that TNF or cachectin treatment of preadipocyte cultures does not affect cell growth or viability as determined by cell counting and by 3 H-thymidine incorporation, as well as by trypan blue exclusion and the clonal growth assay of Ham, R. G., et al., Cell CultureMethods for Molecular and Cellular Biology, Barnes, N.Y. (1984) 1:3-21.
  • the results show that either cachectin or TNF suppresses the total amount of adipose inducible mRNA available in adipocytes or preadipocytes, without influencing negatively the remainder of the cell's metabolism.
  • Example 2 cells cultured and treated as in Example 1 were chilled to 4° C. and the media were aspirated and washed with phosphate buffered saline.
  • Hypotonic buffer (20 mM Tris/HCl, pH 8, 4 mM MgCl 2 , 6 mM CaCl 2 , 0.5 mM dithiothreitol) was added to the plates.
  • 1 ml of lysis buffer (0.6M sucrose, 0.2% NP 40, and 0.5 mM dithiothreitol) was added and the cells were scraped from the tissue culture dishes.
  • nuclei were pelleted at 500 g, washed once in resuspension buffer, repelleted, and then resuspended in buffer containing 0.4 mM each of ATP, CTP, GTP, 10% glycerol, and 10 ⁇ g/ml BSA.
  • the nuclei were incubated with an ⁇ -32P-UTP (600 Ci/mmol) at a concentration of 2 ⁇ Ci/ml for 40 min at 45° with gentle shaking.
  • ⁇ -actin cDNA clone was a gift of P. Gunning and L. Kedes.
  • results are shown in the righthand column of FIG. 2.
  • the four data points are preconfluence (PC). 4 hours after TNF addition, 24 hours after TNF addition, and "control", which represents TAl untreated with TNF.
  • Columns A and E are respectively the ⁇ -actin and pEMBL comparison dot probes;
  • column 28 represents the clone 28 dot probe.
  • FIG. 3 shows that maximal expression of GPD, or clones 1 or 47 is severely inhibited after the first 6 hours in the presence of TNF. Results for cachectin are similar.
  • RNA hybridizable to this clone diminished quickly after TNf administration.
  • 12 ⁇ g total RNA was brought to a final concentration of 2.2M formaldehyde, 30% formamide, 10 mM NaH 2 PO 4 , pH 7 and heated for 15 min at 56°.
  • Samples were electrophoresed in a 1% agarose formaldehyde gel with a final concentration of 2.2M formaldehyde, 20 mM MOPS, pH 7.0, 5.0 mM sodium acetate and 1 mM EDTA.
  • FIGS. 4B and 4A represent untreated and TNF-treated cells, respectively.
  • the oil red O stain has almost entirely disappeared from the cells of FIG. 4A.
  • the L-929 assay system is an improved convenient in vitro assay which permits rapid measurement of TNF activity. Its degree of correlation with the invivo tumor necrosis assay of Carswell is, at present, unknown; however, as it utilizes murine tumor cells specifically, the correlation is expected to be high.
  • the protein designated lymphotoxin in EPO publication no. 0100641 also gives activity in this assay.
  • the assay is similar in concept to that disclosed in U.S. Pat. No. 4,457,916 which used murine L-M cells and methylene blue staining. However, the L-929 assay has been shown to correlate (for HL-60-derived TNF) with human tumor cell line cytotoxicity.
  • L-929 cells are prepared overnight as monolayers in microtiter plates.
  • the test samples are diluted 2-fold across the plate, UV irradiated, and then added onto the prepared cell monolayers.
  • the culture media in the wells are then brought to 1 ⁇ g/ml actinomycin D.
  • the plates are allowed to incubate 18 hr at 37° C. and the plates are scored visually under the microscope.
  • Each well is given a 25, 50, 75 or 100% mark signifying the extent of cell death in the well.
  • One unit of TNF activity is defined as the reciprocal of the dilution at which 50% killing occurs.
  • this assay was developed that monitors the release of 35 S labeled peptides from prelabeled cells, when treated with the test sample and actinomycin D.
  • This version of the assay can be used to quantitate potency, e.g., to evaluate the relative potency of oocyte translated material. Briefly, actively growing L-929 cultures are labeled with 35 S methionine (20 ⁇ Ci/ml) for 3 hr in methionine-free media supplemented with 2% dialyzed fetal calf serum.
  • the cells are then washed and plated into 96 well plates, incubated overnight, and treated the next day with 2-fold dilutions of test samples and 1 ⁇ g/ml actinomycin D.
  • the cultures were then incubated at 37° C. for 18 hr. 100 ⁇ l supernatant aliquots from each well were then transferred onto another 96 well plate, acid (TCA) precipitated, and harvested onto glass fiber filters.
  • TCA acid
  • the filters were washed with 95% ethanol, dried and counted.
  • An NP 40 detergent control is included in every assay to measure maximum release of radioactivity from the cells.
  • the percent 35 S release is then calculated by the ratio of the difference in count between the treated cells and untreated controls divided by the difference between NP 40 treated cells and untreated controls, i.e., by the ratio: ##EQU1## Higher TNF potency results in higher values of this ratio.
  • TNF is successful in suppressing the metabolism of adipose cells. This activity is also demonstrable for cachectin; see Torti, F. M., et al. (supra). Accordingly, TNF prevents the expression of genes responsible for producing enzymes important in storing fat and is useful in controlling weight.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

A method for controlling weight by suppressing the normal metabolism of adipose tissue is disclosed. Administration of tumor necrosis factor (TNF) or a pharmaceutical composition containing it results in suppression of anabolism of adipose cells.

Description

REFERENCE TO GOVERNMENT GRANT
This invention was made with Government support under grant no. GM 25821, awarded by the National Institutes of Health. The Government has certain rights in this invention.
CROSS-REFERENCE TO RELATED APPLICATION
This application is a continuation-in-part of copending U.S. patent application Ser. No. 730,367, filed May 2, 1985.
TECHNICAL FIELD
The invention relates to control of lipid metabolism using tumor necrosis factor (TNF). Specifically, the invention relates to a method of controlling obesity by administration of TNF.
BACKGROUND ART
Control of obesity has become a problem in developed cultures, despite the specter of hunger that often dominates portions of the less technologically advanced world. The litany of less-than-successful approaches which have been used in an attempt to control excess body weight is undoubtedly familar to most. Solutions range from reduced food consumption to often indiscriminate use of pharmaceuticals designed primarily for other purposes, but which appear to have a side effect of somewhat murky mechanism to result in weight loss. In short, there remain millions of people who wish to reduce their body weight without deprivation and without risking unpleasant and perhaps unhealthy side effects.
Of course, not all weight loss is necessarily desirable. Indeed, the weight loss attributable to the chronic catabolic state, referred to as cachexia, developed in the course of infections and malignancies is a handicap to recovery and is often directly fatal. In general, however, cachexia is thought to be a normal response to infection, and is undesirable only when permitted to proceed without proper control. The catabolism is characterized by a net breakdown of lipids in adipose cells, and it has been surmised that this undesirable balance is at least in part a result of failure of the cells to synthesize adequate amounts of lipogenic enzymes.
A crude protein extract from the media of endotoxin-stimulated macrophages, designated "cachectin", has been shown to induce indicia of cachexia in tissue culture (Torti, F., et al., Science (1985) 229: 867-869), and antibodies raised against it have been shown to protect mice from some of the effects of E. coli lipopolysaccharide endotoxin (Beutler, B., et al., ibid, pp. 869-871).
It has now been shown that tumor necrosis factor (TNF) specifically alters the characteristics of adipocytes in tissue culture in a manner thought to model at least part of the cachexia process. Later work has indicated that cachectin and native TNF, which was originally obtained from the sera of endotoxin-treated mice, and which is now available in recombinant form, have the same primary structure. Accordingly, TNF is a useful material for control of weight by stimulating this catabolic reaction under controlled conditions and under circumstances where such stimulation is desirable. It is also possible to control undesirable weight loss by neutralizing TNF.
DISCLOSURE OF THE INVENTION
The invention provides a defined pharmaceutical, tumor necrosis factor (TNF), which is capable of stimulating the physiological state associated with cachexia. The control of weight in adipose persons is thereby effected using a material which simulates a natural physiological response to infection under conditions where such simulation can be regulated and controlled.
Accordingly, in one aspect, the invention relates to a method for controlling obesity by administration of TNF. In another aspect the invention relates to controlling cachexia in cancer or infectious disease patients by providing antibodies to neutralize TNF. In other aspects, the invention relates to pharmaceutical compositions useful for weight reduction which contain TNF as an active ingredient, and to immunoglobulin compositions useful in neutralizing TNF.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows the nucleotide sequence encoding human mature TNF and the deduced amino acid sequence.
FIG. 2 shows the accumulation of adipose-inducible mRNAs in maturing adipose cells and the regulation of synthesis of such mRNA in the presence and absence of TNF.
FIG. 3 shows the depression of expression of adipose stimulated genes by TNF.
FIGS. 4A and 4B are photographs of mature adipose cells with and without TNF.
MODES OF CARRYING OUT THE INVENTION
A. Definitions
As used herein, "tumor necrosis factor" (TNF) refers to an amino acid sequence typified by that shown in FIG. 1, which is capable of selective cytotoxicity against tumor cells. The sequence of FIG. 1 is derived from a human cDNA, but TNF encoded by other mammalian species may exhibit the required activity as well. Retrieval and deduction of this sequence is described in detail in U.S. Ser. No. 760,661, filed July 30, 1985, assigned to the same assignee, and incorporated herein by reference. (See also, Wang, et al., Science (1985) 228:149.)
A TNF amino acid sequence, to fit the definition herein, must be active in the in vitro cytotoxicity assay based on the continuous murine connective tissue cell line L-929 as described hereinbelow. It is recognized that this definition of TNF activity is not precisely the same as that set forth in the disclosure coining this term by Carswell, et al. Proc Natl Acad Sci (USA) (1975) 72:3666. However, this activity as confirmed by the in vitro cytotoxicity assay against human tumor cells provides sufficient assurance of utility that qualification as a tumor necrosis factor for humans using this assay is justified; cytotoxicity against L-929 generalizes to human tumors. It is also expected that there is a substantial overlap between factors active in the specified cytotoxicity assay and the in vivo assay outlined by Carswell.
The TNF protein of the invention, depending on the pH of its environment if suspended or in solution, or of its environment when crystallized or precipitated if in solid form, may be in the form of pharmaceutically acceptable salts or may be in neutral form. The free amino groups of the protein are, of course, capable of forming acid addition salts with, for example, inorganic acids such as hydrochloric, phosphoric, or sulfuric acid; or with organic acids such as, for example, acetic glycolic, succinic, or mandelic acid. The free carboxyl groups are capable of forming salts with bases, including inorganic bases such as sodium, potassium, or calcium hydroxides, and such organic bases as piperidine, glucosamine, trimethylamine, chloine, and caffeine. In addition, the protein may be modified by combination with other biological materials such as lipids and saccharides, or by side chain modification such as acetylation of amino groups, phosphorylation of hydroxyl side chains, or oxidation of sulfhydryl groups. All of these modifications are included within the scope of the definition, so long as the TNF activity is retained.
Finally, it is understood that the primary amino acid sequence shown in FIG. 1 is only illustrative and that similar sequences result in proteins which have substantially equivalent or enhanced activity as compared to that set forth in FIG. 1. These modifications may be deliberate, for example, as obtained through site-directed mutagenesis, or may be accidental such as those obtained through mutation in hosts which are TNF producers. All of these modifications are included as long as TNF activity, as above-defined, is retained.
For example, it has been found that a mutein lacking the first four amino acids at the N-terminus of the sequence shown in FIG. 1 (Val-Arg-Ser-Ser) has a specific activity several fold higher than the TNF of the structure shown. (See U.S. Ser. No. 760,661, filed July 30, 1985, cited above). Accordingly, the definition of TNF specifically includes this truncated form. In addition, muteins lacking the N-terminal ten or less amino acids have similarly enhanced activity, and it appears that deletions of up to 10 amino acids from the N-terminus do not destroy, but, in fact, enhance biological activity.
Therefore, the definition of TNF herein specifically includes proteins having an amino acid sequence substantially equivalent to that shown in FIG. 1, but lacking 1-10 of the amino acids at the N-terminal sequence as shown in that figure.
U.S. Ser. No. 698,939, filed Feb. 7, 1985, assigned to the herein assignee and incorporated herein by reference, discloses cysteine-depleted muteins of the TNF of FIG. 1. In general, neutral amino acid replacements of the crysteine at position 69 or 101 or both result in active TNF proteins. Neutral amino acid replacements include ala, ser, val and the like, preferably ser. These muteins can also be modified to obtain truncated forms which retain TNF activity and may have enhanced specific activity in vitro and in vivo.
As to notation, for convenience, when particular peptides are specified, the protein having the amino acid sequence numbered 1-157 in FIG. 1 will be used as a reference and designated, perhaps arbitrarily, mTNF (mature TNF). All other specific proteins having homology with mTNF and showing TNF biological activity will be referred to as "muteins" of mTNF and will be denoted as to their differences from mTNF using the numbering of residues shown in the figure. For example, muteins which have substitutions for cysteine at position 69 will be denoted using the substituted residue and the position number, e.g., peptides having a serine in place of the cysteine at position 69 are designated ser69 TNF. Muteins which lack, for example, three N-terminal amino acids as compared to the protein shown in FIG. 1 will be designated ∇3TNF. Where both of the foregoing alterations are made, the mutein is designated ∇3ser69 TNF.
Particularly preferred embodiments of the TNF of the inventions include ∇2ser69 TNF, ∇2ser101 TNF, ∇2ser69 ser101 TNF, and the corresponding ∇3, ∇4, ∇5, ∇6, ∇7, ∇8, ∇9 and ∇10 cysteine depleted muteins. Particularly preferred are ∇8ser69 TNF, ∇8ser101 TNF, ∇8ser69 ser101 and ∇4ser69 TNF, ∇4ser101 TNF and ∇4ser69 ser101 TNF.
Not all muteins of mTNF are recombinantly or deliberately produced. Indeed, native HL-60 cell secreted TNF has minor differences from mTNF in the known portions of the primary structure, although both proteins exhibit TNF activity. Specifically, the deduced sequence of FIG. 1 has an additional pair of serine residues following the serine at position 3 as compared to HL-60 derived TNF before resuming the homology shown between positions 4-12 of the HL-60 derived protein and positions 6-14 of the deduced sequence. In addition, positions 13 and 14 of the HL-60 derived protein are Val-Ser; the corresponding positions 15 and 16 of the deduced sequence are His-Val.
B. Modes of Preparation
The TNF useful in the method of the invention can most conveniently be prepared using recombinant methods. Detailed descriptions of ways to produce recombinant TNF are set forth in U.S. Ser. No. 661,026 Oct. 15, 1984), Ser. No. 670,360 (Sept. 11, 1984), Ser. No. 698,939 (Feb. 7, 1985), Ser. No. 730,696 (May 2, 1985), and Ser. No. 760,661 (supra), all incorporated by reference. In this regard, various DNA sequences enconding TNF have been deposited with the American Type Culture Collection, Rockville, MD. These DNA sequences include those contained in pE4 which harbors the human cDNA insert (ATCC #39894); pAW711 which is an expression vector suitable for procaryotes encoding the TNF of FIG. 1 (ATCC #39918) and pAW736, an expression vector encoding the ∇4 mutein of mTNF (ATCC #53092). Vectors suitable for expression of other TNF muteins (∇10, ∇9, ∇6, ∇7, and ∇8) are deposited as ATCC Nos. 53161, 53162, 53163, 53164, and 53165, respectively.
In addition to recombinantly produced materials, the TNF may be extracted from natural source such as human or other mammalian tissues, or from human or other mammalian derived cell lines. The source of the protein is, of course, immaterial to the practice of the invention except as it may affect specific dosage levels and administration regimes required.
C. Formulaion and Mode of Administration
To effect the desired lipid mobilization resulting from administration of TNF, the active ingredient may be formulated using a variety of acceptable excipients as is known in the art. Typically, the TNF will be administered by injection, either intraperitoneally or intravenously. However, by suitable formulations, it may be possible to obtain compositions which may be topically or orally administered, or which may be capable of transmission across mucous membranes.
Depending on the nature of the composition and the mode of administration, the compositions may be in solid or liquid form. For solid compositions, conventional carriers include, for example, phamaceutical grades of mannitol, lactose, starch, talcum, cellulose, magnesium carbonate and the like. The TNF may be formulated as a suppository using, for example, polyalkylene glycols as carrier. Liquid compositions can be prepared by dissolving or dispersing the TNF and optional adjuvants in a carrier such as, for example, water, saline, aqueous dextrose, and so forth. If desired, the composition may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and the like. Actual methods of preparing dosage forms are known or will be apparent to those of the art. The composition will, in any event, contain a quantity of TNF in an amount effective to effect the desired lipid mobilization.
A preferred method of administration of this protein is by injection, most commonly intraperitoneally or intravenously. The injectable can be prepared either as a liquid solution or a suspension or in a solid form susceptible to reconstitution.
The dosage and mode of administration will, of course, depend on the level of lipid mobilization desired, the nature of the subject, and the judgment of the medical practitioner. In general, however, effective dosages are in the range of 0.02-2 μg of TNF per kg of body weight per day for so long as administration is required. This range represents, of course, a broad estimate as the above-listed factors are of great importance in determining optimal dosage levels.
In that aspect of the invention which relates to neutralization of TNF in cachexic patients, a preferred embodiment to neutralize the activity of TNF employs polyclonal or monoclonal antibodies. These neutralizing antibodies are preferably used as adjunctive treatment with other cytotoxic drugs in cancer patients. For example, the patient might be administered methotrexate to kill the tumor followed by the antibody preparation to neutralize TNF to prevent weight loss.
Polyclonal antibodies are prepared to TNF using conventional procedures by injecting purified TNF into a convenient host, such as rabbits or mice, and harvesting high titer sera. Preparation of monoclonal compositions generally follows the now well known procedure of Kohler and Milstein.
D. Example
The following examples are intended to illustrate, but not limit the invention.
Preparation A: Preparation of Cachectin
Cachectin was used as a control in the following examples. It is prepared from the conditioned media of endotoxin stimulated macrophages, as described by Kawakami, M., et al., Proc Natl Acad Sci (USA) (1982) 79:912; Pekala, P. H., et al., (ibid 1983) 80:2743.
Example 1 Effect of TNF on Adipose-Inducible mRNA Levels
An in vitro model for the study of the abnormal metabolism of cachexia has been developed using a stable adipogenic cell line (TAl) as disclosed in Chapman, A. B., J Biol Chem (1984) 259:15548-15555. These cells develop an adipocyte morphology approximately 3 days after confluence in tissue culture monolayers, and show, in parallel with this morphology, the expression of several genes. Certain proteins and enzymes are present only in differentiated adipocytes and are not present or are present only at undetectable levels in the undifferentiated precursors. These proteins are considered to be products of "adipose-inducible genes" and of their intermediate mRNAs.
Genes for which expression is first evident after differentiation have been identified. They are designated clones 1, 10, 28, 47, and GPD (Chapman, A. B., et al., supra). Their expression is apparently due to transcriptional activation.
Therefore, one measure of the impact of compounds on adipocytes is their effect on the levels of adipose-inducible mRNA.
In this assay, the above stable adipogenic cell line, TAl, was cultured from preconfluence to 24 days after confluence. TNF, prepared by culturing pAW711 transformed E. coli and inducing the expression of TNF as described in Ser. No. 760,661 (supra) was added to the cell cultures from preconfluence to 6 days thereafter.
In more detail, TAl cells, which are derived from 5-azacytidine treatment of 10 T1/2C18 cells (Resnikof, C., et al., Cancer Res (1973) 33:3231-3238; Taylor, S. M., et al., Cell (1979) 17:771-779) were grown in Eagle's basal medium supplemented with 10% heat inactivated fetal calf serum. 10-6 M dexamethasone was present in media for the first 3 days after confluence, and 5 μg/ml bovine insulin for the first 6 days after confluence.
Recombinant TNF (10-30 ng/ml) was first added to preadipocyte cultures 2 days prior to confluence. (Addition of this amount inhibits 90% of lipoprotein lipase activity in cultured adipocytes.) Cells were fed with resupplementation of TNF at day 0 (confluence) and at day 3. Cells were harvested at day 6.
Total RNA was isolated by the method of Chirgwin, J. M., et al., Biochemistry (1979) 18:5294-5299, and applied to nitrocellulose in a dot-blot apparatus (BRL). Nick translated cDNA clones of adipose-inducible genes (designated clones 1, 10, 47, and glycerol phosphate dehydrogenase (GDH)) were used to probe the filters under hybridization conditions described in Chapman, A. B., et al., J Biol Chem (1984) (supra). Filters were washed and exposed to XAR 5 film at -70° C. with an intensifying screen.
The results are shown in the left column of FIG. 2, labeled "Steady State". It is clear from these results that TNF treatment prevents accumulation of adipose-inducible mRNA. These results are comparable to those obtained when 10 μl/ml cachectin is used in place of TNF.
Lipid accumulation was also completely inhibited by cachectin or TNF, and TAl cell cultures treated with these compounds have been maintained for as long as 23 days without the appearance of neutral lipid evident by oil red O staining. However, on removal of cachectin or TNF from the medium, adipocyte morphology returns, as does the expression of adipose-inducible genes. It was also shown that TNF or cachectin treatment of preadipocyte cultures does not affect cell growth or viability as determined by cell counting and by 3 H-thymidine incorporation, as well as by trypan blue exclusion and the clonal growth assay of Ham, R. G., et al., Cell CultureMethods for Molecular and Cellular Biology, Barnes, N.Y. (1984) 1:3-21.
Accordingly, the results show that either cachectin or TNF suppresses the total amount of adipose inducible mRNA available in adipocytes or preadipocytes, without influencing negatively the remainder of the cell's metabolism.
Example 2 Inhibition of Transcription
That mRNA accumulation inhibition is transcriptionally regulated was shown by nuclear transcription assays performed as described by Vannice, et al., Proc Natl Acad Sci (USA) (1984) 81:4241-4245 and by Israel, J., et al., J Biol Chem (1984) 259:5400-5402, as modified by Knight, et al. (submitted) for adipose cells.
At each data point, cells cultured and treated as in Example 1 were chilled to 4° C. and the media were aspirated and washed with phosphate buffered saline. One ml of hypotonic buffer (20 mM Tris/HCl, pH 8, 4 mM MgCl2, 6 mM CaCl2, 0.5 mM dithiothreitol) was added to the plates. After 5 min, 1 ml of lysis buffer (0.6M sucrose, 0.2% NP 40, and 0.5 mM dithiothreitol) was added and the cells were scraped from the tissue culture dishes. After Dounce homogenization, nuclei were pelleted at 500 g, washed once in resuspension buffer, repelleted, and then resuspended in buffer containing 0.4 mM each of ATP, CTP, GTP, 10% glycerol, and 10 μg/ml BSA. The nuclei were incubated with an α-32P-UTP (600 Ci/mmol) at a concentration of 2 μCi/ml for 40 min at 45° with gentle shaking.
RNA was harvested from the nuclei as described by Smith, et al., Cell (1978) 15:615-626 as modified by Knight, et al. (supra) and hybridized to linearized cDNAs which had been applied to nitrocellulose filters and baked for 2 hr at 80° C. in a vacuum oven. Filter prehybridization and hybridization conditions were those of Friedman, R. L., et al., Cell (1984) 38:745-755, and hybridizations were performed for 3 days at 42° C. with about 15×106 cpm per reaction mixture applied in 150 μl volume to dots of linearized single-stranded adipose cDNAs for clone 28, and the cDNA encoding β-actin and the non-corresponding neutral plasmid pEMBL were used as comparisons. The β-actin cDNA clone was a gift of P. Gunning and L. Kedes.
The results are shown in the righthand column of FIG. 2. The four data points are preconfluence (PC). 4 hours after TNF addition, 24 hours after TNF addition, and "control", which represents TAl untreated with TNF. Columns A and E are respectively the β-actin and pEMBL comparison dot probes; column 28 represents the clone 28 dot probe. These results show that the transcription system contained in the nucleus is suppressed with respect to the coding sequences for clone 28, which is characteristic of adipose cells, but unaffected with respect to the encoding sequences for actin. Similar results were obtained when the TAl cells were treated with 10 μl/ml cachectin. Therefore, TNF, like cachectin, operates directly on the genome to suppress transcription of adipose-related sequences, but does not disrupt other gene function.
Example 3 Effect of TNF on Mature Adipose Cells
Since in adult mammals, adipocytes undergo little or no proliferation, it was desirable to study the effect of TNF on mature adipose cells. A sample of 10-30 ng/ml of TNF was added to day 6 adipocyte TAl cultures differentiated as described in Example 1. Total RNA was isolated from the cells at various times after TNF exposure and applied to nitrocellulose with a dot blot apparatus. Filters were probed with cDNAs for GPD, clone 47, and clone 1, washed, autoradiographed, and scanned using a Hoeffer GS300 densitometer attached to a reporting integrator. The points obtained were normalized for differences in amount of applied RNA using a cDNA probe made to total cellular RNA. FIG. 3 shows that maximal expression of GPD, or clones 1 or 47 is severely inhibited after the first 6 hours in the presence of TNF. Results for cachectin are similar.
Northern blots probed with clone 47 at various times after the TNF treatment showed that the RNA hybridizable to this clone diminished quickly after TNf administration. For these analyses, 12 μg total RNA was brought to a final concentration of 2.2M formaldehyde, 30% formamide, 10 mM NaH2 PO4, pH 7 and heated for 15 min at 56°. Samples were electrophoresed in a 1% agarose formaldehyde gel with a final concentration of 2.2M formaldehyde, 20 mM MOPS, pH 7.0, 5.0 mM sodium acetate and 1 mM EDTA. Gels were washed in distilled water for 3 min, followed by two 30 min washes in 10 mM NaPO4, pH 7.4 and 1 mM EDTA before transferring to cellulose, and hybridization with linearized cDNA from clone 47. Again, cachectin-treated cells showed similar results.
It was also shown that after 4-6 days of TNF exposure, most cells lose their neutral lipid. When 10-30 ng/ml TNF is added, 70%-80% of cells are laden with large lipid droplets; 6 days later less than 10% of cells have identifiable triglycerides on oil red O stains. These results are shown in FIGS. 4B and 4A, which represent untreated and TNF-treated cells, respectively. The oil red O stain has almost entirely disappeared from the cells of FIG. 4A.
Comparison of these results with those shown in FIG. 3 shows that alterations in adipose specific RNAs due to TNF treatment occur more rapidly than lipid mobilization. By 6-24 hr after addition of TNF to mature TAl lymphocytes, more than a 90% decrease in said RNAs is observed; several days are required for this to be reflected in a decrease in lipid content.
Addendum: Cytotoxic Assay Procedure
Definition of a protein as TNF depends on its activity in the L-929 assay. This assay is therefore described here.
The L-929 assay system is an improved convenient in vitro assay which permits rapid measurement of TNF activity. Its degree of correlation with the invivo tumor necrosis assay of Carswell is, at present, unknown; however, as it utilizes murine tumor cells specifically, the correlation is expected to be high. The protein designated lymphotoxin in EPO publication no. 0100641 also gives activity in this assay. The assay is similar in concept to that disclosed in U.S. Pat. No. 4,457,916 which used murine L-M cells and methylene blue staining. However, the L-929 assay has been shown to correlate (for HL-60-derived TNF) with human tumor cell line cytotoxicity.
In the L-929 assay system herein, L-929 cells are prepared overnight as monolayers in microtiter plates. The test samples are diluted 2-fold across the plate, UV irradiated, and then added onto the prepared cell monolayers. The culture media in the wells are then brought to 1 μg/ml actinomycin D. The plates are allowed to incubate 18 hr at 37° C. and the plates are scored visually under the microscope. Each well is given a 25, 50, 75 or 100% mark signifying the extent of cell death in the well. One unit of TNF activity is defined as the reciprocal of the dilution at which 50% killing occurs.
In addition, a more sensitive version of this assay was developed that monitors the release of 35 S labeled peptides from prelabeled cells, when treated with the test sample and actinomycin D. This version of the assay can be used to quantitate potency, e.g., to evaluate the relative potency of oocyte translated material. Briefly, actively growing L-929 cultures are labeled with 35 S methionine (20 μCi/ml) for 3 hr in methionine-free media supplemented with 2% dialyzed fetal calf serum. The cells are then washed and plated into 96 well plates, incubated overnight, and treated the next day with 2-fold dilutions of test samples and 1 μg/ml actinomycin D. The cultures were then incubated at 37° C. for 18 hr. 100 μl supernatant aliquots from each well were then transferred onto another 96 well plate, acid (TCA) precipitated, and harvested onto glass fiber filters. The filters were washed with 95% ethanol, dried and counted. An NP40 detergent control is included in every assay to measure maximum release of radioactivity from the cells. The percent 35 S release is then calculated by the ratio of the difference in count between the treated cells and untreated controls divided by the difference between NP40 treated cells and untreated controls, i.e., by the ratio: ##EQU1## Higher TNF potency results in higher values of this ratio.
Summary
The foregoing examples indicate that TNF is successful in suppressing the metabolism of adipose cells. This activity is also demonstrable for cachectin; see Torti, F. M., et al. (supra). Accordingly, TNF prevents the expression of genes responsible for producing enzymes important in storing fat and is useful in controlling weight.

Claims (12)

We claim:
1. A method of effecting weight reduction by administering to a subject in need of such treatment an amount of tumor necrosis factor (TNF) effective to cause said weight reduction or a pharmaceutical composition containing said amount,
wherein said TNF is the mTNF having the amino acid sequence shown in FIG. 1 or a mutein thereof,
wherein said mutein is selected from the group consisting of ∇1TNF, ∇2-, ∇3- . . . through ∇10TNF;
ser69 TNF, ∇1ser69 TNF, ∇2ser69 TNF . . . through ∇10ser69 TNF;
ser101 TNF, ∇1ser101 TFN, ∇2ser101 TNF . . . through ∇10ser101 TNF;
ser69 ser101 TNF, ∇1ser69 ser101 TNF, ∇2ser69 ser101 TNF . . . through ∇10ser69 ser101 TNF; and
a mutein having the sequence of amino acids 21-157 of FIG. 1 with the N-terminal sequence Val-Arg-Ser-Arg-Thr-Pro-Ser-Asp-Lys-Pro-Val-Ala-Val-Ser-Val-Ala-Asn-Pro.
2. A method of suppressing adipose cell metabolism which comprises administering to a subject in need of such treatment an amount of TNF effective to suppress adipose cell metabolism or a pharmaceutical composition containing said amount,
wherein said TNF is the mTNF having the amino acid sequence shown in FIG. 1 or a mutein thereof,
wherein said mutein is selected from the group consisting of ∇1TNF, ∇2-, ∇3- . . . through ∇10TNF;
∇1ser69 TNF, ∇2ser69 TNF . . . through ∇10ser69 TNF;
ser101 TNF, ∇1ser101 TNF, ∇2ser101 TNF . . . ∇10ser101 TNF;
ser69 ser101 TNF, ∇1ser69 ser101 TNF, ∇2ser69 ser101 TNF . . . ∇10ser69 ser101 TNF; and
a mutein having the sequence of amino acids 21-157 of FIG. 1, with the N-terminal sequence Val-Arg-Ser-Arg-Thr-Pro-Ser-Asp-Lys-Pro-Val-Ala-Val-Ser-Val-Ala-Asn-Pro.
3. The method of claim 1 wherein the TNF has an amino acid sequence substantially equivalent to that of FIG. 1 (mTNF).
4. The method of claim 1 wherein the TNF is recombinant TNF.
5. The method of claim 1 wherein the TNF is selected from the group consisting of ∇1TNF, ∇2-, ∇3-, . . . ∇10TNF.
6. The method of claim 1 wherein the TNF is selected from the group consisting of ser69 TNF, ∇1ser69 TNF, ∇2ser69 TNF, ∇3-, . . . ∇10ser69 TNF.
7. The method of claim 1 wherein the TNF is selected from the group consisting of ser101 TNF, ∇1ser101 TNF, ∇2ser101 TNF, ∇3-, . . . ∇10ser101 TNF.
8. The method of claim 1 wherein the TNF is selected from the group consisting of ser69 ser101 TNF, ∇1ser69 ser101 TNF, ∇2ser69 ser101 TNF, ∇3-, . . . ∇10ser69 ser101 TNF.
9. The method of claim 1 wherein the TNF is selected from the group consisting of ∇8ser69 TNF, ∇8ser101 TNF, and ∇8ser69 ser101 TMF.
10. The method of claim 1 wherein the TNF is selected from the group consisting of ∇4ser69 TNF, ∇4ser101 TNF, and ∇4ser69 ser101 TNF.
11. The method of claim 1 wherein the TNF has the N-terminal sequence:
Val-Arg-Ser-Arg-Thr-Pro-Ser-Asp-Lys-Pro-Val-Ala-Val-Ser-Val-Ala-Asn-Pro.
12. A method of controlling undesirable weight loss which comprises administering to a subject in need of such control a neutralizing amount of antibodies specific for the TNF of claim 1 or a pharmaceutical composition thereof.
US06/801,989 1985-05-02 1985-11-26 Use of tumor necrosis factor as a weight regulator Expired - Fee Related US4684623A (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US06/801,989 US4684623A (en) 1985-05-02 1985-11-26 Use of tumor necrosis factor as a weight regulator
PCT/US1986/000952 WO1986006280A1 (en) 1985-05-02 1986-05-01 Use of tumor necrosis factor as a weight regulator
EP86903084A EP0226603A1 (en) 1985-05-02 1986-05-01 Use of tumor necrosis factor as a weight regulator
AU58628/86A AU5862886A (en) 1985-05-02 1986-05-01 Use of tumor necrosis factor as a weight regulator
FI865373A FI865373A0 (en) 1985-05-02 1986-12-31 ANALYZING AV TUMOERNEKROSFAKTOR SOM VIKTREGULATOR.
NO870008A NO870008L (en) 1985-05-02 1987-01-02 USE OF TUMOR CROSS FACTOR AS WEIGHT CONTROLLER.
DK001887A DK1887A (en) 1985-05-02 1987-01-02 USE OF TUMOR CROSS FACTOR AS A WEIGHT CONTROLLER

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73036785A 1985-05-02 1985-05-02
US06/801,989 US4684623A (en) 1985-05-02 1985-11-26 Use of tumor necrosis factor as a weight regulator

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US73036785A Continuation-In-Part 1985-05-02 1985-05-02

Publications (1)

Publication Number Publication Date
US4684623A true US4684623A (en) 1987-08-04

Family

ID=27112039

Family Applications (1)

Application Number Title Priority Date Filing Date
US06/801,989 Expired - Fee Related US4684623A (en) 1985-05-02 1985-11-26 Use of tumor necrosis factor as a weight regulator

Country Status (6)

Country Link
US (1) US4684623A (en)
EP (1) EP0226603A1 (en)
AU (1) AU5862886A (en)
DK (1) DK1887A (en)
FI (1) FI865373A0 (en)
WO (1) WO1986006280A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4822776A (en) * 1981-09-08 1989-04-18 The Rockefeller University Lipoprotein lipase suppression by endotoxin-induced mediator (shock assay)
EP0330521A2 (en) * 1988-02-26 1989-08-30 STOLLE RESEARCH & DEVELOPMENT CORPORATION Antihypertensive hyperimmune milk
WO1990006365A1 (en) * 1988-11-30 1990-06-14 The Beth Israel Hospital Association Recombinant proteins with adipsin and complement d activities
US5102658A (en) * 1985-07-19 1992-04-07 National Research Development Corporation Antibody treatment for adiposity
US5322699A (en) * 1991-02-04 1994-06-21 The Rockefeller University Leukocyte-derived CR3 modulator, integrin modulating factor-1 (IMF-1)
WO1995005849A1 (en) * 1993-08-26 1995-03-02 Mouritsen & Elsner A/S Inducing antibody response against self-proteins with the aid of foreign t-cell epitopes
US5560908A (en) * 1993-01-22 1996-10-01 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Therapeutic agent for NIDDM
US5658803A (en) * 1988-07-18 1997-08-19 Chiron Corporation Monoclonal antibodies reactive with cachectin
US5733742A (en) * 1993-06-03 1998-03-31 Therapeutic Antibodies Inc. Production of antibody fragments from whole blood
US6090923A (en) * 1984-12-20 2000-07-18 Yeda Research And Development Co. Ltd. Murine monoclonal antibody binding TNFα
US6309640B1 (en) 1981-09-08 2001-10-30 The Rockefeller University Lipoprotein lipase suppression by endotoxin-induced mediator (shock assay)
US6354297B1 (en) * 1998-04-16 2002-03-12 The Uniformed Services University Of The Health Sciences Method and device for destroying fat cells by induction of programmed cell death
US6419927B1 (en) 1981-09-08 2002-07-16 Anthony Cerami Method for reducing adverse effects of a human 70kDa mediator which results from endotoxin stimulation of macrophages
US20030022856A1 (en) * 2001-01-30 2003-01-30 The Regents Of The University Of Michigan Method for sustained release local delivery of drugs for ablation of unwanted tissue
US20050222040A1 (en) * 2004-04-05 2005-10-06 Blm Group, Inc. Vertebrate peptide modulators of lipid metabolism
US7285269B2 (en) 2002-12-02 2007-10-23 Amgen Fremont, Inc. Antibodies directed to tumor necrosis factor

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE114673T1 (en) * 1985-08-16 1994-12-15 Univ Rockefeller ANABOLIC ACTIVITY MODULATOR AND ITS USES.
US6180549B1 (en) * 1998-09-10 2001-01-30 The B. F. Goodrich Company Modified zeolites and methods of making thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4457916A (en) * 1982-08-31 1984-07-03 Asahi Kasei Kogyo Kabushiki Kaisha Method for stabilizing Tumor Necrosis Factor and a stable aqueous solution or powder containing the same

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Beutler, B. et al., Science, 229: 869 871. *
Beutler, B. et al., Science, 229: 869-871.
J. Exp. Med., vol. 154 (1981), 631 639. *
J. Exp. Med., vol. 154 (1981), 631-639.
Proc. Nat l. Acad. Sci., 79, 912 916 (1982). *
Proc. Nat'l. Acad. Sci., 79, 912-916 (1982).
Torti, F. M. et al., Science, 229: 867 869. *
Torti, F. M. et al., Science, 229: 867-869.

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4822776A (en) * 1981-09-08 1989-04-18 The Rockefeller University Lipoprotein lipase suppression by endotoxin-induced mediator (shock assay)
US6419927B1 (en) 1981-09-08 2002-07-16 Anthony Cerami Method for reducing adverse effects of a human 70kDa mediator which results from endotoxin stimulation of macrophages
US6309640B1 (en) 1981-09-08 2001-10-30 The Rockefeller University Lipoprotein lipase suppression by endotoxin-induced mediator (shock assay)
US6090923A (en) * 1984-12-20 2000-07-18 Yeda Research And Development Co. Ltd. Murine monoclonal antibody binding TNFα
US5102658A (en) * 1985-07-19 1992-04-07 National Research Development Corporation Antibody treatment for adiposity
EP0330521A3 (en) * 1988-02-26 1990-10-31 Stolle Research And Development Corporation Antihypertensive hyperimmune milk
EP0330521A2 (en) * 1988-02-26 1989-08-30 STOLLE RESEARCH & DEVELOPMENT CORPORATION Antihypertensive hyperimmune milk
US5658803A (en) * 1988-07-18 1997-08-19 Chiron Corporation Monoclonal antibodies reactive with cachectin
WO1990006365A1 (en) * 1988-11-30 1990-06-14 The Beth Israel Hospital Association Recombinant proteins with adipsin and complement d activities
US5322699A (en) * 1991-02-04 1994-06-21 The Rockefeller University Leukocyte-derived CR3 modulator, integrin modulating factor-1 (IMF-1)
US5560908A (en) * 1993-01-22 1996-10-01 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Therapeutic agent for NIDDM
US5733742A (en) * 1993-06-03 1998-03-31 Therapeutic Antibodies Inc. Production of antibody fragments from whole blood
US6193969B1 (en) 1993-06-03 2001-02-27 Protherics Inc. Antibody fragments in therapy
WO1995005849A1 (en) * 1993-08-26 1995-03-02 Mouritsen & Elsner A/S Inducing antibody response against self-proteins with the aid of foreign t-cell epitopes
US6354297B1 (en) * 1998-04-16 2002-03-12 The Uniformed Services University Of The Health Sciences Method and device for destroying fat cells by induction of programmed cell death
US20030022856A1 (en) * 2001-01-30 2003-01-30 The Regents Of The University Of Michigan Method for sustained release local delivery of drugs for ablation of unwanted tissue
US7285269B2 (en) 2002-12-02 2007-10-23 Amgen Fremont, Inc. Antibodies directed to tumor necrosis factor
US8101178B2 (en) 2002-12-02 2012-01-24 Amgen Fremont Inc. Antibodies directed to tumor necrosis factor and uses thereof
US20050222040A1 (en) * 2004-04-05 2005-10-06 Blm Group, Inc. Vertebrate peptide modulators of lipid metabolism

Also Published As

Publication number Publication date
FI865373A (en) 1986-12-31
EP0226603A1 (en) 1987-07-01
WO1986006280A1 (en) 1986-11-06
DK1887A (en) 1987-03-02
DK1887D0 (en) 1987-01-02
FI865373A0 (en) 1986-12-31
AU5862886A (en) 1986-11-18

Similar Documents

Publication Publication Date Title
US4684623A (en) Use of tumor necrosis factor as a weight regulator
Pfefferkorn et al. Inhibition of growth of Toxoplasma gondii in cultured fibroblasts by human recombinant gamma interferon
Torti et al. A macrophage factor inhibits adipocyte gene expression: an in vitro model of cachexia
Shanley et al. Regulation of chemokine expression by IL-10 in lung inflammation
JP3320774B2 (en) Pharmaceutical composition
Wolf et al. B94, a primary response gene inducible by tumor necrosis factor-alpha, is expressed in developing hematopoietic tissues and the sperm acrosome.
JPH09505721A (en) Interleukin 6 receptor antagonist
US20030171253A1 (en) Methods and compositions relating to modulation of A20
JP2003527311A (en) Treatment of fibrosis by antagonizing IL-13 and IL-receptor chains
JP2007039464A (en) LYMPHOTOXIN-beta, LYMPHOTOXIN-beta COMPLEX, PHARMACEUTICAL PREPARATION AND THERAPEUTIC USE THEREOF
JP2007332145A (en) Chemokine binding protein and method for using thereof
EP0567575B1 (en) Cytokine-induced protein, tsg-6, dna coding therefor and uses thereof
DE69912925T2 (en) VACCINE COMPOSITION AGAINST HERPES SIMPLEX VIRUS AND METHODS FOR USE
HU218598B (en) Process for producing pharmaceutical compositions containing interleukin-10, for treating neoplastic disease
Hangai et al. Increased cytokine gene expression in rat retina following transient ischemia
Myers et al. Discriminatory effects of protein kinase inhibitors and calcium ionophore on endothelial ICAM-1 induction
ES2415331T3 (en) Derivatives of the gamma chain of the IL-2 receptor, its preparation and use
KR960015199B1 (en) Bcrfi protein as inhibitor of interferon-ñò
Holstad et al. A transcriptional inhibitor of TNF-α prevents diabetes induced by multiple low-dose streptozotocin injections in mice
NO337024B1 (en) Use of cytokine-1 IL-1F7b which can bind IL-18BP, for the preparation of a medicament for the treatment or prevention of inflammatory diseases, and a vector encoding said cytokine and use of said vector
Pritts et al. Peroxisome proliferator-activated receptor-γ ligand inhibition of RANTES production by human endometriotic stromal cells is mediated through an upstream promoter element
US5157106A (en) N-terminal deletions of lymphotoxin, their preparation and use
Sveinbjørnsson et al. Cytotoxic effect of cytokines on murine colon carcinoma cells involves TNF-mediated apoptosis
EP1466619A2 (en) Method for modulating cell apoptosis
JP2863265B2 (en) Interleukin 1 inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST.;ASSIGNORS:RINGOLD, GORDON M.;TORTI, FRANK M.;REEL/FRAME:004511/0080

Effective date: 19860106

Owner name: CETUS CORPORATION, 1400 FIFTY-THIRD STREET, EMERYV

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST.;ASSIGNORS:LARRICK, JAMES W.;LIN, LEO S.;MARK, DAVID F.;REEL/FRAME:004511/0078

Effective date: 19860103

CC Certificate of correction
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Expired due to failure to pay maintenance fee

Effective date: 19910804